Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Stem Cell ; 26(2): 251-261.e8, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-31956039

RESUMO

Vascular smooth muscle cells (VSMCs) can be derived in large numbers from human induced pluripotent stem cells (hiPSCs) for producing tissue-engineered vascular grafts (TEVGs). However, hiPSC-derived TEVGs are hampered by low mechanical strength and significant radial dilation after implantation. Here, we report generation of hiPSC-derived TEVGs with mechanical strength comparable to native vessels used in arterial bypass grafts by utilizing biodegradable scaffolds, incremental pulsatile stretching, and optimal culture conditions. Following implantation into a rat aortic model, hiPSC-derived TEVGs show excellent patency without luminal dilation and effectively maintain mechanical and contractile function. This study provides a foundation for future production of non-immunogenic, cellularized hiPSC-derived TEVGs composed of allogenic vascular cells, potentially serving needs to a considerable number of patients whose dysfunctional vascular cells preclude TEVG generation via other methods.


Assuntos
Prótese Vascular , Células-Tronco Pluripotentes Induzidas , Humanos , Miócitos de Músculo Liso , Engenharia Tecidual
2.
Acta Biomater ; 102: 220-230, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31634626

RESUMO

Single ventricle heart defects (SVDs) are congenital disorders that result in a variety of complications, including increased ventricular mechanical strain and mixing of oxygenated and deoxygenated blood, leading to heart failure without surgical intervention. Corrective surgery for SVDs are traditionally handled by the Fontan procedure, requiring a vascular conduit for completion. Although effective, current conduits are limited by their inability to aid in pumping blood into the pulmonary circulation. In this report, we propose an innovative and versatile design strategy for a tissue engineered pulsatile conduit (TEPC) to aid circulation through the pulmonary system by producing contractile force. Several design strategies were tested for production of a functional TEPC. Ultimately, we found that porcine extracellular matrix (ECM)-based engineered heart tissue (EHT) composed of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and primary cardiac fibroblasts (HCF) wrapped around decellularized human umbilical artery (HUA) made an efficacious basal TEPC. Importantly, the TEPCs showed effective electrical and mechanical function. Initial pressure readings from our TEPC in vitro (0.68 mmHg) displayed efficient electrical conductivity enabling them to follow electrical pacing up to a 2 Hz frequency. This work represents a proof of principle study for our current TEPC design strategy. Refinement and optimization of this promising TEPC design will lay the groundwork for testing the construct's therapeutic potential in the future. Together this work represents a progressive step toward developing an improved treatment for SVD patients. STATEMENT OF SIGNIFICANCE: Single Ventricle Cardiac defects (SVD) are a form of congenital disorder with a morbid prognosis without surgical intervention. These patients are treated through the Fontan procedure which requires vascular conduits to complete. Fontan conduits have been traditionally made from stable or biodegradable materials with no pumping activity. Here, we propose a tissue engineered pulsatile conduit (TEPC) for use in Fontan circulation to alleviate excess strain in SVD patients. In contrast to previous strategies for making a pulsatile Fontan conduit, we employ a modular design strategy that allows for the optimization of each component individually to make a standalone tissue. This work sets the foundation for an in vitro, trainable human induced pluripotent stem cell based TEPC.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Miócitos Cardíacos/fisiologia , Engenharia Tecidual/métodos , Artérias Umbilicais/fisiologia , Animais , Diferenciação Celular/fisiologia , Colágeno Tipo I/química , Matriz Extracelular/fisiologia , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Ácido Poliglicólico/química , Estudo de Prova de Conceito , Suínos , Alicerces Teciduais/química
3.
Biomaterials ; 212: 28-38, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31102854

RESUMO

Metallic stents cause vascular wall damage with subsequent smooth muscle cell (SMC) proliferation, neointimal hyperplasia, and treatment failure. To combat in-stent restenosis, drug-eluting stents (DES) delivering mTOR inhibitors such as sirolimus or everolimus have become standard for coronary stenting. However, the relatively non-specific action of mTOR inhibitors prevents efficient endothelium recovery and mandates dual antiplatelet therapy to prevent thrombosis. Unfortunately, long-term dual antiplatelet therapy leads to increased risk of bleeding/stroke and, paradoxically, myocardial infarction. Here, we took advantage of the fact that nitric oxide (NO) increases Fas receptors on the SMC surface. Fas forms a death-inducing complex upon binding to Fas ligand (FasL), while endothelial cells (ECs) are relatively resistant to this pathway. Selected doses of FasL and NO donor synergistically increased SMC apoptosis and inhibited SMC growth more potently than did everolimus or sirolimus, while having no significant effect on EC viability and proliferation. This differential effect was corroborated in ex vivo pig coronaries, where the neointimal formation was inhibited by the drug combination, but endothelial viability was retained. We also deployed FasL-NO donor-releasing ethylene-vinyl acetate copolymer (EVAc)-coated stents into pig coronary arteries, and cultured them in perfusion bioreactors for one week. FasL and NO donor, released from the stent coating, killed SMCs close to the stent struts, even in the presence of flow rates mimicking those of native arteries. Thus, the FasL-NO donor-combination has a potential to prevent intimal hyperplasia and in-stent restenosis, without harming endothelial restoration, and hence may be a superior drug delivery strategy for DES.


Assuntos
Células Endoteliais/citologia , Proteína Ligante Fas/farmacologia , Miócitos de Músculo Liso/citologia , Óxido Nítrico/farmacologia , Sirolimo/farmacologia , Animais , Reatores Biológicos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Vasos Coronários/citologia , Células Endoteliais/efeitos dos fármacos , Everolimo/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Miócitos de Músculo Liso/efeitos dos fármacos , Compostos Nitrosos/farmacologia , Polímeros/química , Suínos
4.
Cell Transplant ; 27(9): 1375-1389, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30095004

RESUMO

OBJECTIVE: Invasive coronary interventions can fail due to intimal hyperplasia and restenosis. Endothelial cell (EC) seeding to the vessel lumen, accelerating re-endothelialization, or local release of mTOR pathway inhibitors have helped reduce intimal hyperplasia after vessel injury. While animal models are powerful tools, they are complex and expensive, and not always reflective of human physiology. Therefore, we developed an in vitro 3D vascular model validating previous in vivo animal models and utilizing isolated human arteries to study vascular remodeling after injury. APPROACH: We utilized a bioreactor that enables the control of intramural pressure and shear stress in vessel conduits to investigate the vascular response in both rat and human arteries to intraluminal injury. RESULTS: Culturing rat aorta segments in vitro, we show that vigorous removal of luminal ECs results in vessel injury, causing medial proliferation by Day-4 and neointima formation, with the observation of SCA1+ cells (stem cell antigen-1) in the intima by Day-7, in the absence of flow. Conversely, when endothelial-denuded rat aortae and human umbilical arteries were subjected to arterial shear stress, pre-seeding with human umbilical ECs decreased the number and proliferation of smooth muscle cell (SMC) significantly in the media of both rat and human vessels. CONCLUSION: Our bioreactor system provides a novel platform for correlating ex vivo findings with vascular outcomes in vivo. The present in vitro human arterial injury model can be helpful in the study of EC-SMC interactions and vascular remodeling, by allowing for the separation of mechanical, cellular, and soluble factors.


Assuntos
Artérias/lesões , Artérias/patologia , Perfusão/instrumentação , Remodelação Vascular , Animais , Aorta/lesões , Aorta/patologia , Reatores Biológicos , Desenho de Equipamento , Células Endoteliais da Veia Umbilical Humana , Humanos , Ratos , Ratos Sprague-Dawley , Técnicas de Cultura de Tecidos , Artérias Umbilicais/lesões , Artérias Umbilicais/patologia
5.
Ann Thorac Surg ; 105(6): 1809-1818, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29510094

RESUMO

BACKGROUND: Synthetic graft materials are commonly used for shunts and cardiovascular reconstruction in neonates, but are prone to thrombosis and scarring. The umbilical vein is a potential source of autologous, endothelialized tissue for neonatal shunts and tissue reconstruction, but requires preservation before implantation. METHODS: Umbilical cords were collected in UW solution with antibiotics at 4°C until dissection. Umbilical vein segments were tested for burst pressure before and after 2 weeks of preservation. Umbilical veins segments were preserved under static or flow conditions at 4°C in UW solution with 5% human plasma lysate for 7 days. Veins were evaluated with histopathology, scanning electron microscopy, and platelet adhesion testing. RESULTS: Umbilical veins have no difference in burst pressure at harvest (n = 16) compared with 2 weeks of preservation (n = 11; 431 ± 229 versus 438 ± 244 mm Hg). After 1 week, static and flow-preserved veins showed viability of the vessel segments with endothelium staining positive for CD31, von Willebrand factor, and endothelial nitric oxide synthase. Scanning electron microscopy demonstrated preservation of normal endothelial morphology and flow alignment in the flow-preserved samples compared with cobblestone endothelial appearance and some endothelial cell loss in the static samples. Static samples had significantly more platelet adhesion than flow-preserved samples did. CONCLUSIONS: Umbilical veins have adequate burst strength to function at neonatal systemic pressures. Preservation under flow conditions demonstrated normal endothelial and overall vascular morphology with less platelet adhesion compared with static samples. Preserved autologous umbilical veins are potential source for endothelialized shunts or cardiovascular repair tissue for neonates.


Assuntos
Endotélio Vascular/diagnóstico por imagem , Endotélio Vascular/fisiologia , Soluções para Preservação de Órgãos/química , Procedimentos de Cirurgia Plástica/métodos , Preservação de Tecido/métodos , Veias Umbilicais/transplante , Biópsia por Agulha , Procedimentos Cirúrgicos Cardíacos/métodos , Feminino , Humanos , Imuno-Histoquímica , Recém-Nascido , Masculino , Microscopia Eletrônica de Varredura/métodos , Sensibilidade e Especificidade , Coleta de Tecidos e Órgãos/métodos , Transplante Autólogo/métodos , Veias Umbilicais/cirurgia , Veias Umbilicais/ultraestrutura
6.
Stem Cells Dev ; 27(7): 441-455, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29415620

RESUMO

Pluripotent stem cell-derived endothelial cells (ECs) have great potential to be used in vascular therapy or tissue engineering. It is also much desired to obtain arterial or venous ECs for specific applications. Factors that are critical for the proper arterial or venous differentiation from pluripotent stem cells still need to be understood. Here, we aim at investigating this problem deeper by examining neuropilin-1 (Nrp1), an early arterial marker that may be critical for arterial cell fate commitment. Using murine embryonic stem cells as the model system, this study investigates the neuropilin-1 (Nrp1) expression during the differentiation of pluripotent stem cells toward a vascular progenitor population. We hypothesize that Nrp1, an early arterial marker present in a developing embryo, may be more responsive when further induced in vitro toward an arterial fate. We developed a two-step differentiation approach that yielded a large percentage of Nrp1+ vascular progenitor cells (VPCs) and investigated their potential to become arterial ECs. We have defined the culture parameters that contribute greatly to the emergence of Nrp1+ VPCs: certain soluble factors, especially Wnt and BMP4, early cell-cell contact, and hypoxia. Subsequent isolation of this population demonstrated a highly proliferative and network-forming behavior. The Nrp1+ VPCs exhibited increased gene expression of several Notch pathway-related arterial markers compared with Nrp1- VPCs. Most importantly, Nrp1+ VPCs demonstrated a dramatically greater response to hemodynamic stimuli by upregulating many arterial markers whereas Nrp1- VPCs have very little response. Surprisingly, these differences between Nrp1+ and Nrp1- VPCs are not evident with vascular endothelial growth factor (VEGF) treatment. Our data suggest that Nrp1+ VPCs may serve as the arterial progenitor by enhanced response to hemodynamic flow but not to VEGF, whereas Nrp1- VPCs lack the plasticity to become arterial ECs. The findings of this research indicate that Nrp1+ VPCs in the murine model act as an important step in the arterial differentiation process.


Assuntos
Diferenciação Celular/genética , Células Endoteliais/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Neuropilina-1/genética , Células-Tronco Pluripotentes/metabolismo , Animais , Artérias/citologia , Artérias/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Expressão Gênica/efeitos dos fármacos , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Neuropilina-1/metabolismo , Células-Tronco Pluripotentes/citologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
7.
Biomaterials ; 147: 116-132, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28942128

RESUMO

Development of autologous tissue-engineered vascular constructs using vascular smooth muscle cells (VSMCs) derived from human induced pluripotent stem cells (iPSCs) holds great potential in treating patients with vascular disease. However, preclinical, large animal iPSC-based cellular and tissue models are required to evaluate safety and efficacy prior to clinical application. Herein, swine iPSC (siPSC) lines were established by introducing doxycycline-inducible reprogramming factors into fetal fibroblasts from a line of inbred Massachusetts General Hospital miniature swine that accept tissue and organ transplants without immunosuppression within the line. Highly enriched, functional VSMCs were derived from siPSCs based on addition of ascorbic acid and inactivation of reprogramming factor via doxycycline withdrawal. Moreover, siPSC-VSMCs seeded onto biodegradable polyglycolic acid (PGA) scaffolds readily formed vascular tissues, which were implanted subcutaneously into immunodeficient mice and showed further maturation revealed by expression of the mature VSMC marker, smooth muscle myosin heavy chain. Finally, using a robust cellular self-assembly approach, we developed 3D scaffold-free tissue rings from siPSC-VSMCs that showed comparable mechanical properties and contractile function to those developed from swine primary VSMCs. These engineered vascular constructs, prepared from doxycycline-inducible inbred siPSCs, offer new opportunities for preclinical investigation of autologous human iPSC-based vascular tissues for patient treatment.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Engenharia Tecidual/métodos , Animais , Ácido Ascórbico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Vasos Coronários/fisiologia , Células Endoteliais , Fibroblastos/citologia , Células HEK293 , Humanos , Masculino , Camundongos , Contração Muscular , Músculo Liso Vascular/fisiologia , Ácido Poliglicólico/química , Suínos , Alicerces Teciduais
8.
Tissue Eng Part A ; 22(17-18): 1086-97, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27520928

RESUMO

Here we report the creation of a novel tracheal construct in the form of an engineered, acellular tissue-stent biocomposite trachea (TSBT). Allogeneic or xenogeneic smooth muscle cells are cultured on polyglycolic acid polymer-metal stent scaffold leading to the formation of a tissue comprising cells, their deposited collagenous matrix, and the stent material. Thorough decellularization then produces a final acellular tubular construct. Engineered TSBTs were tested as end-to-end tracheal replacements in 11 rats and 3 nonhuman primates. Over a period of 8 weeks, no instances of airway perforation, infection, stent migration, or erosion were observed. Histological analyses reveal that the patent implants remodel adaptively with native host cells, including formation of connective tissue in the tracheal wall and formation of a confluent, columnar epithelium in the graft lumen, although some instances of airway stenosis were observed. Overall, TSBTs resisted collapse and compression that often limit the function of other decellularized tracheal replacements, and additionally do not require any cells from the intended recipient. Such engineered TSBTs represent a model for future efforts in tracheal regeneration.


Assuntos
Bioprótese , Teste de Materiais , Stents , Engenharia Tecidual , Alicerces Teciduais/química , Traqueia , Animais , Bovinos , Chlorocebus aethiops , Humanos , Ratos
9.
Arterioscler Thromb Vasc Biol ; 36(9): 1847-53, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27386939

RESUMO

OBJECTIVE: It is widely accepted that the presence of a glycosaminoglycan-rich glycocalyx is essential for endothelialized vasculature health; in fact, a damaged or impaired glycocalyx has been demonstrated in many vascular diseases. Currently, there are no methods that characterize glycocalyx functionality, thus limiting investigators' ability to assess the role of the glycocalyx in vascular health. APPROACH AND RESULTS: We have developed novel, easy-to-use, in vitro assays that directly quantify live endothelialized surface's functional heparin weights and their anticoagulant capacity to inactivate Factor Xa and thrombin. Using our assays, we characterized 2 commonly used vascular models: native rat aorta and cultured human umbilical vein endothelial cell monolayer. We determined heparin contents to be ≈10 000 ng/cm(2) on the native aorta and ≈10-fold lower on cultured human umbilical vein endothelial cells. Interestingly, human umbilical vein endothelial cells demonstrated a 5-fold lower anticoagulation capacity in inactivating both Factor Xa and thrombin relative to native aortas. We verified the validity and accuracy of the novel assays developed in this work using liquid chromatography-mass spectrometry analysis. CONCLUSIONS: Our assays are of high relevance in the vascular community because they can be used to establish the antithrombogenic capacity of many different types of surfaces such as vascular grafts and transplants. This work will also advance the capacity for glycocalyx-targeting therapeutics development to treat damaged vasculatures.


Assuntos
Aorta Torácica/metabolismo , Bioensaio/métodos , Coagulação Sanguínea , Fator Xa/metabolismo , Glicocálix/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Trombina/metabolismo , Animais , Antitrombinas/metabolismo , Aorta Torácica/ultraestrutura , Células Cultivadas , Cromatografia Líquida , Glicocálix/ultraestrutura , Heparina/metabolismo , Heparitina Sulfato/metabolismo , Células Endoteliais da Veia Umbilical Humana/ultraestrutura , Masculino , Espectrometria de Massas , Microscopia Eletrônica de Transmissão , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Fatores de Tempo
10.
Biomaterials ; 102: 120-9, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27336184

RESUMO

Derivation of functional vascular smooth muscle cells (VSMCs) from human induced pluripotent stem cells (hiPSCs) to generate tissue-engineered blood vessels (TEBVs) holds great potential in treating patients with vascular diseases. Herein, hiPSCs were differentiated into alpha-smooth muscle actin (α-SMA) and calponin-positive VSMCs, which were seeded onto polymer scaffolds in bioreactors for vascular tissue growth. A functional TEBV with abundant collagenous matrix and sound mechanics resulted, which contained cells largely positive for α-SMA and smooth muscle myosin heavy chain (SM-MHC). Moreover, when hiPSC-derived TEBV segments were implanted into nude rats as abdominal aorta interposition grafts, they remained unruptured and patent with active vascular remodeling, and showed no evidence of teratoma formation during a 2-week proof-of-principle study. Our studies represent the development of the first implantable TEBVs based on hiPSCs, and pave the way for developing autologous or allogeneic grafts for clinical use in patients with vascular disease.


Assuntos
Prótese Vascular , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos de Músculo Liso/citologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Ratos Nus , Alicerces Teciduais/química
11.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L221-8, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25416381

RESUMO

Cellular therapy via direct intratracheal delivery has gained interest as a novel therapeutic strategy for treating various pulmonary diseases including cystic fibrosis lung disease. However, concerns such as insufficient cell engraftment in lungs and lack of large animal model data remain to be resolved. This study aimed to establish a simple method for evaluating cell retention in lungs and to develop reproducible approaches for efficient cell delivery into mouse and pig lungs. Human lung epithelial cells including normal human bronchial/tracheal epithelial (NHBE) cells and human lung epithelial cell line A549 were infected with pSicoR-green fluorescent protein (GFP) lentivirus. GFP-labeled NHBE cells were delivered via a modified intratracheal cell instillation method into the lungs of C57BL/6J mice. Two days following cell delivery, GFP ELISA-based assay revealed a substantial cell-retention efficiency (10.48 ± 2.86%, n = 7) in mouse lungs preinjured with 2% polidocanol. When GFP-labeled A549 cells were transplanted into Yorkshire pig lungs with a tracheal intubation fiberscope, a robust initial cell attachment (22.32% efficiency) was observed at 24 h. In addition, a lentiviral vector was developed to induce the overexpression and apical localization of cystic fibrosis transmembrane conductance regulator (CFTR)-GFP fusion proteins in NHBE cells as a means of ex vivo CFTR gene transfer in nonprogenitor (relatively differentiated) lung epithelial cells. These results have demonstrated the convenience and efficiency of direct delivery of exogenous epithelial cells to lungs in mouse and pig models and provided important background for future preclinical evaluation of intratracheal cell transplantation to treat lung diseases.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Epiteliais/transplante , Lesão Pulmonar/terapia , Mucosa Respiratória/citologia , Mucosa Respiratória/transplante , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/biossíntese , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Pulmão/citologia , Pulmão/metabolismo , Lesão Pulmonar/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Polidocanol , Polietilenoglicóis , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Suínos
12.
Curr Opin Chem Eng ; 3: 68-74, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24533306

RESUMO

Tissue and organ replacement is required when there are no alternative therapies available. Although vascular tissue engineering was originally developed to meet the clinical demands of small-diameter vascular conduits as bypass grafts, it has evolved into a highly advanced field where perfusable vasculatures are generated for implantation. Herein, we review several cutting-edge techniques that have led to implantable human blood vessels in clinical trials, the novel approaches that build complex perfusable microvascular networks in functional tissues, the use of stem cells to generate endothelial cells for vascularization, as well as the challenges in bringing vascular tissue engineering technologies into the clinics.

13.
Tissue Eng Part A ; 20(9-10): 1499-507, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24320793

RESUMO

Tissue-engineered small-diameter vascular grafts have been developed as a promising alternative to native veins or arteries for replacement therapy. However, there is still a crucial need to improve the current approaches to render the tissue-engineered blood vessels more favorable for clinical applications. A completely biological blood vessel (3-mm inner diameter) was constructed by culturing a 50:50 mixture of bovine smooth muscle cells (SMCs) with neonatal human dermal fibroblasts in fibrin gels. After 30 days of culture under pulsatile stretching, the engineered blood vessels demonstrated an average burst pressure of 913.3±150.1 mmHg (n=6), a suture retention (53.3±15.4 g) that is suitable for implantation, and a compliance (3.1%±2.5% per 100 mmHg) that is comparable to native vessels. These engineered grafts contained circumferentially aligned collagen fibers, microfibrils and elastic fibers, and differentiated SMCs, mimicking a native artery. These promising mechanical and biochemical properties were achieved in a very short culture time of 30 days, suggesting the potential of co-culturing SMCs with fibroblasts in fibrin gels to generate functional small-diameter vascular grafts for vascular reconstruction surgery.


Assuntos
Prótese Vascular , Vasos Sanguíneos/crescimento & desenvolvimento , Fibrina/química , Fibroblastos/fisiologia , Miócitos de Músculo Liso/fisiologia , Engenharia Tecidual/instrumentação , Alicerces Teciduais , Animais , Vasos Sanguíneos/citologia , Bovinos , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/citologia , Humanos , Miócitos de Músculo Liso/citologia , Desenho de Prótese , Engenharia Tecidual/métodos
14.
J Clin Invest ; 123(11): 4950-62, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24135142

RESUMO

The use of induced pluripotent stem cells (iPSCs) has been postulated to be the most effective strategy for developing patient-specific respiratory epithelial cells, which may be valuable for lung-related cell therapy and lung tissue engineering. We generated a relatively homogeneous population of alveolar epithelial type II (AETII) and type I (AETI) cells from human iPSCs that had phenotypic properties similar to those of mature human AETII and AETI cells. We used these cells to explore whether lung tissue can be regenerated in vitro. Consistent with an AETII phenotype, we found that up to 97% of cells were positive for surfactant protein C, 95% for mucin-1, 93% for surfactant protein B, and 89% for the epithelial marker CD54. Additionally, exposing induced AETII to a Wnt/ß-catenin inhibitor (IWR-1) changed the iPSC-AETII-like phenotype to a predominantly AETI-like phenotype. We found that of induced AET1 cells, more than 90% were positive for type I markers, T1α, and caveolin-1. Acellular lung matrices were prepared from whole rat or human adult lungs treated with decellularization reagents, followed by seeding these matrices with alveolar cells derived from human iPSCs. Under appropriate culture conditions, these progenitor cells adhered to and proliferated within the 3D lung tissue scaffold and displayed markers of differentiated pulmonary epithelium.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Alvéolos Pulmonares/citologia , Células Epiteliais Alveolares/classificação , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/metabolismo , Animais , Biomarcadores/metabolismo , Adesão Celular , Diferenciação Celular , Proliferação de Células , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Mucina-1/metabolismo , Alvéolos Pulmonares/metabolismo , Proteína B Associada a Surfactante Pulmonar/metabolismo , Proteína C Associada a Surfactante Pulmonar/metabolismo , Ratos , Engenharia Tecidual , Alicerces Teciduais
15.
Tissue Eng Part A ; 17(9-10): 1191-200, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21143045

RESUMO

Functional connective tissues have been developed using tissue engineering approach by seeding cells on biodegradable scaffolds such as polyglycolic acid (PGA). However, a major drawback of tissue engineering approaches that utilize synthetic polymers is the persistence of polymer remnants in engineered tissues at the end of culture. Such polymer fragments may significantly degrade tissue mechanics and stimulate local inflammatory responses in vivo. In this study, several polymeric materials with a range of degradation profiles were developed and evaluated for their potential applications in construction of collagen matrix-rich tissues, particularly tissue-engineered blood vessels. The degradation characteristics of these polymers were compared as were their characteristics vis-à-vis cell adhesion and proliferation, collagen synthesis, and ability to support growth of engineered vessels. Under aqueous conditions at 37°C, Polymer I (comprising 84% glycolide and 16% trimethylene carbonate [TMC]) had a similar degradation profile to PGA, Polymer II (comprising 84% glycolide, 14% TMC, and 2% polyethylene succinate) degradedly more slowly, but Polymer III (comprising 87% glycolide, 7% TMC, and 6% polyethylene glycol) had a more extensive degradation as compared to PGA. All polymers supported cell proliferation, but Polymer III improved collagen production and engineered vessel mechanics as compared with PGA. In addition, more slowly degrading polymers were associated with poorer final vessel mechanics. These results suggest that polymers that degrade more quickly during tissue culture actually result in improved engineered tissue mechanics, by virtue of decreased disruption of collagenous extracellular matrix.


Assuntos
Implantes Absorvíveis , Prótese Vascular , Teste de Materiais , Polímeros/química , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Suínos
16.
Science ; 329(5991): 538-41, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20576850

RESUMO

Because adult lung tissue has limited regeneration capacity, lung transplantation is the primary therapy for severely damaged lungs. To explore whether lung tissue can be regenerated in vitro, we treated lungs from adult rats using a procedure that removes cellular components but leaves behind a scaffold of extracellular matrix that retains the hierarchical branching structures of airways and vasculature. We then used a bioreactor to culture pulmonary epithelium and vascular endothelium on the acellular lung matrix. The seeded epithelium displayed remarkable hierarchical organization within the matrix, and the seeded endothelial cells efficiently repopulated the vascular compartment. In vitro, the mechanical characteristics of the engineered lungs were similar to those of native lung tissue, and when implanted into rats in vivo for short time intervals (45 to 120 minutes) the engineered lungs participated in gas exchange. Although representing only an initial step toward the ultimate goal of generating fully functional lungs in vitro, these results suggest that repopulation of lung matrix is a viable strategy for lung regeneration.


Assuntos
Matriz Extracelular , Pulmão , Regeneração , Engenharia Tecidual/métodos , Animais , Reatores Biológicos , Detergentes , Células Endoteliais/citologia , Células Endoteliais/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Matriz Extracelular/fisiologia , Humanos , Pulmão/irrigação sanguínea , Pulmão/citologia , Pulmão/fisiologia , Complacência Pulmonar , Transplante de Pulmão , Masculino , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/ultraestrutura , Troca Gasosa Pulmonar , Ratos , Ratos Endogâmicos F344 , Mucosa Respiratória/citologia , Alicerces Teciduais
17.
Cell Transplant ; 19(1): 79-87, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19878625

RESUMO

While advances in regenerative medicine and vascular tissue engineering have been substantial in recent years, important stumbling blocks remain. In particular, the limited life span of differentiated cells that are harvested from elderly human donors is an important limitation in many areas of regenerative medicine. Recently, a mutant of the human telomerase reverse transcriptase enzyme (TERT) was described, which is highly processive and elongates telomeres more rapidly than conventional telomerase. This mutant, called pot1-TERT, is a chimeric fusion between the DNA binding protein pot1 and TERT. Because pot1-TERT is highly processive, it is possible that transient delivery of this transgene to cells that are utilized in regenerative medicine applications may elongate telomeres and extend cellular life span while avoiding risks that are associated with retroviral or lentiviral vectors. In the present study, adenoviral delivery of pot1-TERT resulted in transient reconstitution of telomerase activity in human smooth muscle cells, as demonstrated by telomeric repeat amplification protocol (TRAP). In addition, human engineered vessels that were cultured using pot1-TERT-expressing cells had greater collagen content and somewhat better performance in vivo than control grafts. Hence, transient delivery of pot1-TERT to elderly human cells may be useful for increasing cellular life span and improving the functional characteristics of resultant tissue-engineered constructs.


Assuntos
Vasos Sanguíneos/transplante , Proteínas Recombinantes de Fusão/uso terapêutico , Telomerase/uso terapêutico , Proteínas de Ligação a Telômeros/uso terapêutico , Engenharia Tecidual/métodos , Transfecção/métodos , Adenoviridae/genética , Adulto , Animais , Reatores Biológicos/normas , Vasos Sanguíneos/citologia , Técnicas de Cultura de Células , Células Cultivadas , Senescência Celular/genética , Colágeno/metabolismo , Vetores Genéticos/uso terapêutico , Sobrevivência de Enxerto/genética , Humanos , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/transplante , Ratos , Ratos Nus , Proteínas Recombinantes de Fusão/genética , Complexo Shelterina , Telomerase/genética , Proteínas de Ligação a Telômeros/genética
18.
Tissue Eng Part C Methods ; 16(2): 173-84, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19419244

RESUMO

Decellularization of native tissues is a promising technique with numerous applications in tissue engineering and regenerative medicine. However, there are various limitations of currently available decellularization methods, such as alteration of extracellular matrix mechanics and restricted use on certain tissues. This study was conducted to explore the effect of serum on the decellularization of various types of tissues. Fetal bovine serum-containing cell culture medium endothelial growth media-2 removed DNA but not cellular beta-actin from human umbilical artery after detergent treatment, without compromising the tissue mechanical strength assessed by burst pressure. In addition, the effect of serum-containing endothelial growth media-2 on DNA removal was replicated in other types of tissues such as tissue-engineered vessels and myocardium. Other types of serum, including human serum, were also shown to remove DNA from detergent-pretreated tissues. In conclusion, we describe a novel utilization of serum that may have broad applications in tissue decellularization.


Assuntos
Fracionamento Celular/métodos , Soro/fisiologia , Engenharia Tecidual/métodos , Animais , Fenômenos Biomecânicos , Vasos Sanguíneos/fisiologia , Bovinos , Fracionamento Celular/estatística & dados numéricos , Células Cultivadas , Meios de Cultura/química , Meios de Cultura/farmacologia , DNA/isolamento & purificação , Detergentes/farmacologia , Coração/fisiologia , Humanos , Teste de Materiais , Ratos , Ratos Endogâmicos F344 , Soro/química , Estresse Mecânico , Suínos , Artérias Umbilicais/citologia
19.
Tissue Eng Part A ; 15(9): 2665-76, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19207043

RESUMO

OBJECTIVE: Developing a tissue-engineered small-diameter (<6mm) vascular graft for reconstructive surgery has remained a challenge for the past several decades. This study was conducted to develop a decellularized umbilical artery and to evaluate its composition, endothelial cell compatibility, mechanical properties, and in vivo stability for potential use as a small-diameter vascular graft. METHODS AND RESULTS: Human umbilical arteries were isolated and decellularized by incubation in CHAPS and sodium dodecyl sulfate buffers followed by incubation in endothelial growth media-2. Decellularized umbilical arteries were completely devoid of cellular and nuclear material while retaining the integrity of extracellular collagenous matrix. The mechanical strength of the decellularized umbilical artery as assessed by its burst pressure in vitro showed no significant change from its native form. Decellularized umbilical arteries supported endothelial adherence as indicated by the re-endotheliazation with a monolayer of human umbilical vein endothelial cells. Furthermore, decellularized vessels that were implanted into nude rats as abdominal aorta interposition grafts remained mechanically intact and patent for up to 8 weeks. CONCLUSION: Decellularized human umbilical arteries preserved the extracellular matrix, supported endothelialization, and retained function in vivo for up to 8 weeks. These properties suggest the potential use of decellularized umbilical arteries as small-diameter vascular grafts.


Assuntos
Prótese Vascular , Engenharia Tecidual/métodos , Artérias Umbilicais/citologia , Animais , Implante de Prótese Vascular , Colágeno Tipo IV/metabolismo , DNA/metabolismo , Células Endoteliais/citologia , Matriz Extracelular/metabolismo , Imunofluorescência , Humanos , Hiperplasia , Fenômenos Mecânicos , Perfusão , Ratos , Ratos Nus , Coloração e Rotulagem , Artérias Umbilicais/patologia , Artérias Umbilicais/transplante
20.
J Biol Chem ; 281(46): 34816-25, 2006 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-16982604

RESUMO

Fibronectins are high molecular mass glycoproteins that circulate as soluble molecules in the blood, and are also found in an insoluble, multimeric form in extracellular matrices throughout the body. Soluble fibronectins are polymerized into insoluble extracellular matrix (ECM) fibrils via a cell-dependent process. Recent studies indicate that the interaction of cells with the ECM form of fibronectin promotes actin organization and cell contractility, increases cell growth and migration, and enhances the tensile strength of artificial tissue constructs; ligation of integrins alone is insufficient to trigger these responses. Evidence suggests that the effect of ECM fibronectin on cell function is mediated in part by a matricryptic heparin-binding site within the first III1 repeat (FNIII1). In this study, we localized the heparin-binding activity of FNIII1 to a cluster of basic amino acids, Arg613, Trp614, Arg615, and Lys617. Site-directed mutagenesis of a recombinant fibronectin construct engineered to mimic the ECM form of fibronectin demonstrates that these residues are also critical for stimulating cell spreading and increasing cell proliferation. Cell proliferation has been tightly correlated with cell area. Using integrin- and heparin-binding fibronectin mutants, we found a positive correlation between cell spreading and growth when cells were submaximally spread on ECM protein-coated surfaces at the time of treatment. However, cells maximally spread on vitronectin or fibronectin still responded to the fibronectin matrix mimetic with an increase in growth, indicating that an absolute change in cell area is not required for the increase in cell proliferation induced by the matricryptic site of FNIII1.


Assuntos
Crescimento Celular , Fibronectinas/química , Fibronectinas/metabolismo , Heparina/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proliferação de Células , Tamanho Celular , Matriz Extracelular , Fibroblastos , Humanos , Integrinas/genética , Integrinas/metabolismo , Camundongos , Mutação , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA